Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Metab ; 6(1): 113-126, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38167727

RESUMO

Chronic stress and inflammation are both outcomes and major drivers of many human diseases. Sustained responsiveness despite mitigation suggests a failure to sense resolution of the stressor. Here we show that a proteolytic cleavage event of fatty acid synthase (FASN) activates a global cue for stress resolution in Caenorhabditis elegans. FASN is well established for biosynthesis of the fatty acid palmitate. Our results demonstrate FASN promoting an anti-inflammatory profile apart from palmitate synthesis. Redox-dependent proteolysis of limited amounts of FASN by caspase activates a C-terminal fragment sufficient to downregulate multiple aspects of stress responsiveness, including gene expression, metabolic programs and lipid droplets. The FASN C-terminal fragment signals stress resolution in a cell non-autonomous manner. Consistent with these findings, FASN processing is also seen in well-fed but not fasted male mouse liver. As downregulation of stress responses is critical to health, our findings provide a potential pathway to control diverse aspects of stress responses.


Assuntos
Ácido Graxo Sintases , Ácidos Graxos , Animais , Masculino , Camundongos , Ácido Graxo Sintases/genética , Ácido Graxo Sintases/metabolismo , Palmitatos , Proteólise , Caenorhabditis elegans , Ácido Graxo Sintase Tipo I
2.
JCI Insight ; 8(16)2023 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-37432749

RESUMO

Reactive oxygen species (ROS) are natural products of mitochondrial oxidative metabolism and oxidative protein folding. ROS levels must be well controlled, since elevated ROS has been shown to have deleterious effects on osteoblasts. Moreover, excessive ROS is thought to underlie many of the skeletal phenotypes associated with aging and sex steroid deficiency in mice and humans. The mechanisms by which osteoblasts regulate ROS and how ROS inhibits osteoblasts are not well understood. Here, we demonstrate that de novo glutathione (GSH) biosynthesis is essential in neutralizing ROS and establish a proosteogenic reduction and oxidation reaction (REDOX) environment. Using a multifaceted approach, we demonstrate that reducing GSH biosynthesis led to acute degradation of RUNX2, impaired osteoblast differentiation, and reduced bone formation. Conversely, reducing ROS using catalase enhanced RUNX2 stability and promoted osteoblast differentiation and bone formation when GSH biosynthesis was limited. Highlighting the therapeutic implications of these findings, in utero antioxidant therapy stabilized RUNX2 and improved bone development in the Runx2+/- haplo-insufficient mouse model of human cleidocranial dysplasia. Thus, our data establish RUNX2 as a molecular sensor of the osteoblast REDOX environment and mechanistically clarify how ROS negatively impacts osteoblast differentiation and bone formation.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core , Osteogênese , Camundongos , Humanos , Animais , Osteogênese/genética , Espécies Reativas de Oxigênio , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Oxirredução , Glutationa/metabolismo
3.
Curr Osteoporos Rep ; 20(1): 53-64, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35112289

RESUMO

PURPOSE OF REVIEW: Osteoblasts are responsible for bone matrix production during bone development and homeostasis. Much is known about the transcriptional regulation and signaling pathways governing osteoblast differentiation. However, less is known about how osteoblasts obtain or utilize nutrients to fulfill the energetic demands associated with osteoblast differentiation and bone matrix synthesis. The goal of this review is to highlight and discuss what is known about the role and regulation of bioenergetic metabolism in osteoblasts with a focus on more recent studies. RECENT FINDINGS: Bioenergetic metabolism has emerged as an important regulatory node in osteoblasts. Recent studies have begun to identify the major nutrients and bioenergetic pathways favored by osteoblasts as well as their regulation during differentiation. Here, we highlight how osteoblasts obtain and metabolize glucose, amino acids, and fatty acids to provide energy and other metabolic intermediates. In addition, we highlight the signals that regulate nutrient uptake and metabolism and focus on how energetic metabolism promotes osteoblast differentiation. Bioenergetic metabolism provides energy and other metabolites that are critical for osteoblast differentiation and activity. This knowledge contributes to a more comprehensive understanding of osteoblast biology and may inform novel strategies to modulate osteoblast differentiation and bone anabolism in patients with bone disorders.


Assuntos
Osteoblastos , Osteogênese , Desenvolvimento Ósseo , Diferenciação Celular , Metabolismo Energético/fisiologia , Humanos , Osteoblastos/metabolismo
4.
Sci Rep ; 11(1): 1626, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33452305

RESUMO

Osteoarthritis is a debilitating disease characterized by cartilage degradation and altered cartilage mechanical properties. Furthermore, it is well established that obesity is a primary risk factor for osteoarthritis. The purpose of this study was to investigate the influence of obesity on the mechanical properties of murine knee cartilage. Two-month old wild type mice were fed either a normal diet or a high fat diet for 16 weeks. Atomic force microscopy-based nanoindentation was used to quantify the effective indentation modulus of medial femoral condyle cartilage. Osteoarthritis progression was graded using the OARSI system. Additionally, collagen organization was evaluated with picrosirius red staining imaged using polarized light microscopy. Significant differences between diet groups were assessed using t tests with p < 0.05. Following 16 weeks of a high fat diet, no significant differences in OARSI scoring were detected. However, we detected a significant difference in the effective indentation modulus between diet groups. The reduction in cartilage stiffness is likely the result of disrupted collagen organization in the superficial zone, as indicated by altered birefringence on polarized light microscopy. Collectively, these results suggest obesity is associated with changes in knee cartilage mechanical properties, which may be an early indicator of disease progression.


Assuntos
Cartilagem Articular/metabolismo , Colágeno/metabolismo , Módulo de Elasticidade , Obesidade/patologia , Animais , Cartilagem Articular/patologia , Dieta Hiperlipídica , Modelos Animais de Doenças , Teste de Tolerância a Glucose , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Força Atômica , Obesidade/complicações , Obesidade/metabolismo , Osteoartrite/etiologia , Osteoartrite/metabolismo , Osteoartrite/patologia , Fatores de Transcrição SOX9/metabolismo
5.
Am J Pathol ; 190(8): 1701-1712, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32416098

RESUMO

Interleukin 17A (IL-17A) is critical in the pathogenesis of autoimmune diseases through driving inflammatory cascades. However, the role of IL-17 in osteoarthritis (OA) is not well understood. Tumor necrosis factor-receptor-associated factor 3 (TRAF3) is a receptor proximal negative regulator of IL-17 signaling. It remains unclear whether TRAF3 exerts regulatory effects on cartilage degradation and contributes to the pathogenesis of OA. In this study, we found that TRAF3 notably suppressed IL-17-induced NF-κB and mitogen-activated protein kinase activation and, subsequently, the production of matrix-degrading enzymes. TRAF3 depletion enhanced IL-17 signaling, along with increased matrix-degrading enzyme production. In vivo, cartilage destruction caused by surgery-induced OA was alleviated markedly both in 1l17a-deficient mice and in TRAF3 transgenic mice. In contrast, silencing TRAF3 through adenoviruses worsened cartilage degradation in experimental OA. Moreover, the destructive effect of IL-17 on cartilage was abolished in TRAF3 transgenic mice in an IL-17 intra-articular injection animal model. Similarly, genetic deletion of IL-17 blocked TRAF3 knockdown-mediated promotion of cartilage destruction, suggesting that the protective effect of TRAF3 on cartilage is mediated by its suppression of IL-17 signaling. Collectively, our results suggest that TRAF3 negatively regulates IL-17-mediated cartilage degradation and pathogenesis of OA, and may serve as a potential new therapy target for OA.


Assuntos
Artrite Experimental/metabolismo , Cartilagem Articular/metabolismo , Interleucina-17/metabolismo , Osteoartrite/metabolismo , Transdução de Sinais/fisiologia , Fator 3 Associado a Receptor de TNF/metabolismo , Animais , Artrite Experimental/genética , Artrite Experimental/patologia , Cartilagem Articular/patologia , Condrócitos/metabolismo , Condrócitos/patologia , Camundongos , Camundongos Transgênicos , NF-kappa B/metabolismo , Osteoartrite/genética , Osteoartrite/patologia , Fator 3 Associado a Receptor de TNF/genética
6.
J Bone Miner Res ; 35(10): 2004-2014, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32453500

RESUMO

Skeletal stem/progenitor cells (SSPC) are critical regulators of bone homeostasis by providing a continuous supply of osteoblasts throughout life. In response to inductive signals, SSPC proliferate before osteoblast differentiation. Proliferation requires the duplication of all cellular components before cell division. This imposes a unique biosynthetic requirement for amino acids that can be used for biomass production. Thus, the ability to sense and respond to amino acid availability is likely a major determinant for proliferation. Using a cellular and genetic approach, we demonstrate the amino acid sensor GCN2 is required to support the robust proliferative capacity of SSPC during bone homeostasis. GCN2 ablation results in decreased postnatal bone mass due primarily to reduced osteoblast numbers. Decreased osteoblast numbers is likely attributed to reduced SSPC proliferation as loss of GCN2 specifically affected proliferation in cultured bone marrow stromal cells (BMSCs) without impacting osteoblast differentiation in vitro. Mechanistically, GCN2 regulates proliferation by increasing amino acid uptake downstream of the transcriptional effector ATF4. Collectively, these data suggest amino acid sensing through the GCN2/ATF4 pathway is indispensable for robust SSPC proliferation necessary for bone homeostasis. © 2020 American Society for Bone and Mineral Research.


Assuntos
Proliferação de Células , Osteoblastos/citologia , Proteínas Serina-Treonina Quinases/fisiologia , Células-Tronco/citologia , Animais , Diferenciação Celular , Camundongos
7.
J Mol Cell Biol ; 12(1): 55-70, 2020 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30986855

RESUMO

PIP5k1ß is crucial to the generation of phosphotidylinosotol (4, 5)P2. PIP5k1ß participates in numerous cellular activities, such as B cell and platelet activation, cell phagocytosis and endocytosis, cell apoptosis, and cytoskeletal organization. In the present work, we aimed to examine the function of PIP5k1ß in osteoclastogenesis and osteogenesis to provide promising strategies for osteoporosis prevention and treatment. We discovered that PIP5k1ß deletion in mice resulted in obvious bone loss and that PIP5k1ß was highly expressed during both osteoclast and osteoblast differentiation. Deletion of the gene was found to enhance the proliferation and migration of bone marrow-derived macrophage-like cells to promote osteoclast differentiation. PIP5k1ß-/- osteoclasts exhibited normal cytoskeleton architecture but stronger resorption activity. PIP5k1ß deficiency also promoted activation of mitogen-activated kinase and Akt signaling, enhanced TRAF6 and c-Fos expression, facilitated the expression and nuclear translocation of NFATC1, and upregulated Grb2 expression, thereby accelerating osteoclast differentiation and function. Finally, PIP5k1ß enhanced osteoblast differentiation by upregulating master gene expression through triggering smad1/5/8 signaling. Therefore, PIP5k1ß modulates bone homeostasis and remodeling.


Assuntos
Remodelação Óssea/genética , Diferenciação Celular/genética , Homeostase/genética , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Movimento Celular/genética , Células Cultivadas , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Transdução de Sinais/genética
8.
J Mol Cell Biol ; 12(4): 305-317, 2020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-31065679

RESUMO

Tendinopathy, which is characterized by the ectopic ossification of tendon, is a common disease occurring in certain population, such as athletes that suffer from repetitive tendon strains. However, the molecular mechanism underlying the pathogenesis of tendinopathy caused by the overuse of tendon is still lacking. Here, we found that the mechanosensitive miRNA, miR-337-3p, had lower expression under uniaxial cyclical mechanical loading in tendon-derived stem cells (TDSCs) and negatively controlled chondro-osteogenic differentiation of TDSCs. Importantly, downregulation of miR-337-3p expression was also observed in both rat and human calcified tendons, and overexpressing miR-337-3p in patellar tendons of rat tendinopathy model displayed a robust therapeutic efficiency. Mechanistically, we found that the proinflammatory cytokine interleukin-1ß was the upstream factor of miR-337-3p that bridges the mechanical loading with its downregulation. Furthermore, the target genes of miR-337-3p, NADPH oxidase 4, and insulin receptor substrate 1, activated chondro-osteogenic differentiation of TDSCs through JNK and ERK signaling, respectively. Thus, these findings not only provide novel insight into the molecular mechanisms underlying ectopic ossification in tendinopathy but also highlight the significance of miR-337-3p as a putative therapeutic target for clinic treatment of tendinopathy.


Assuntos
Proteínas Substratos do Receptor de Insulina/metabolismo , MicroRNAs/metabolismo , NADPH Oxidase 4/metabolismo , Ossificação Heterotópica/genética , Células-Tronco/metabolismo , Tendinopatia/genética , Tendões/patologia , Animais , Sequência de Bases , Diferenciação Celular/genética , Condrogênese/genética , Colagenases/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Interleucina-1beta/metabolismo , Masculino , MicroRNAs/genética , Modelos Biológicos , Ossificação Heterotópica/patologia , Osteogênese/genética , Ratos , Ratos Sprague-Dawley , Tendinopatia/patologia , Transcrição Gênica , Suporte de Carga
9.
J Bone Miner Res ; 34(11): 2149-2161, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31356686

RESUMO

MicroRNAs play important roles in osteoporosis and show great potential for diagnosis and therapy of osteoporosis. Previous studies have demonstrated that miR-146a affects osteoblast (OB) and osteoclast (OC) formation. However, these findings have yet to be identified in vivo, and it is unclear whether miR-146a is related to postmenopausal osteoporosis. Here, we demonstrated that miR-146a knockout protects bone loss in mouse model of estrogen-deficient osteoporosis, and miR-146a inhibits OB and OC activities in vitro and in vivo. MiR-146a-/- mice displayed the same bone mass as the wild type (WT) but exhibited a stronger bone turnover than the WT did under normal conditions. Nevertheless, miR-146a-/- mice showed an increase in bone mass after undergoing ovariectomy (OVX) compared with those subjected to sham operation. OC activities were impaired in the miR-146a-/- mice exposed to estrogen deficiency, which was diametrically opposite to the enhanced bone resorption ability of WT. Macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL)/osteoprotegerin (OPG) from a bone microenvironment affect this extraordinary phenomenon. Therefore, our results implicate that miR-146a plays a key role in estrogen deficiency-induced osteoporosis, and the inhibition of this molecule provides skeleton protection. © 2019 American Society for Bone and Mineral Research.


Assuntos
Microambiente Celular/genética , Deleção de Genes , Fator Estimulador de Colônias de Macrófagos/metabolismo , MicroRNAs/genética , Osteoporose , Osteoprotegerina/metabolismo , Ligante RANK/metabolismo , Animais , Camundongos , Camundongos Knockout , MicroRNAs/metabolismo , Osteoporose/genética , Osteoporose/metabolismo , Osteoporose/patologia , Osteoprotegerina/genética , Ovariectomia , Ligante RANK/genética
10.
Cell Metab ; 29(4): 966-978.e4, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30773468

RESUMO

Skeletal stem cells (SSCs) are postulated to provide a continuous supply of osteoblasts throughout life. However, under certain conditions, the SSC population can become incorrectly specified or is not maintained, resulting in reduced osteoblast formation, decreased bone mass, and in severe cases, osteoporosis. Glutamine metabolism has emerged as a critical regulator of many cellular processes in diverse pathologies. The enzyme glutaminase (GLS) deaminates glutamine to form glutamate-the rate-limiting first step in glutamine metabolism. Using genetic and metabolic approaches, we demonstrate GLS and glutamine metabolism are required in SSCs to regulate osteoblast and adipocyte specification and bone formation. Mechanistically, transaminase-dependent α-ketoglutarate production is critical for the proliferation, specification, and differentiation of SSCs. Collectively, these data suggest stimulating GLS activity may provide a therapeutic approach to expand SSCs in aged individuals and enhance osteoblast differentiation and activity to increase bone mass.


Assuntos
Linhagem da Célula , Glutamina/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Feminino , Glutamina/análise , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos
11.
Oncotarget ; 9(4): 4833-4850, 2018 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-29435145

RESUMO

Although the mechanisms of Tumor necrosis factor alpha (TNF-α) on facilitating osteoclast differentiation and bone resorption is well known, the mechanisms behind the suppression of the osteoblast differentiation from mesenchymal stem cells (MSCs) are still poorly understood. In this study, we observed a negative correlation between TNF-α levels and the expression of special AT-rich sequence-binding protein 2 (SATB2), a critical osteoblastogenesis transcription factor, in ovariectomy (OVX)-induced bone loss and IL-1-induced arthritis animal model. We found that TNF-α treatment inhibited mesenchymal cell line C2C12 osteoblast differentiation and sharply decreased BMP2-induced SATB2 expression. Upon TNF-α treatment, the activity of smad1/5/8 was inhibited, by contrast, extracellular signal-regulated kinase-1/2 (ERK1/2) and P38 was increased in C2C12 cells, the inhibitor of ERK1/2 (U0126) was found to abrogate the TNF-α inhibition of SATB2 expression. Furthermore, the NF-κB signaling pathway in C2C12 cells was significantly activated by the treatment of TNF-α, and TNF-α induced NF-κB directly binds to SATB2 promoter to suppress its expression. These results suggest that TNF-α suppresses SATB2 expression through activating NF-κB and MAPK signaling and depressing smad1/5/8 signaling, which contributes to the inhibition of osteoblast differentiation and might be potential therapeutic targets for inflammation-induced bone loss.

12.
J Bone Miner Metab ; 36(6): 648-660, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29234953

RESUMO

Elucidation of the molecular mechanism governing bone marrow mesenchymal stem cell (BMSC) osteogenic differentiation is of great importance for improving the treatment of osteoporosis. TNF-α is a well-known inhibitory factor during osteogenic differentiation of BMSCs. In our experiment, we consistently observed that TNF-α significantly inhibited BMSC osteogenic differentiation, which was partially rescued by BAY 11-7082 (NF-κB inhibitor). In this study, we examined the potential roles of microRNAs (miRNAs) involved in TNF-α-mediated reduction of BMSC osteogenesis. We found that microRNA-23b (miR-23b) was dramatically induced under the stimulation of TNF-α, which was abolished by BAY 11-7082. Similar to the effect of TNF-α, miR-23b agonist (agomir-23b) obviously impaired BMSC osteogenic differentiation in vitro and in vivo. However, agomir-23b had no effect on osteoclast activity. Overexpression of miR-23b significantly reduced runx2, the master transcription factor during osteogenesis, suggesting that miR-23b acts as an endogenous attenuator of runx2 in BMSCs. Mutation of the putative miR-23b binding site in runx2 mRNA blocked miR-23b-mediated repression of the runx2 3' untranslated region (3'UTR) luciferase reporter activity, suggesting that miR-23b directly binds to runx2 3'UTR. Furthermore, infection with Ad-runx2 (adenovirus carrying the entire CDS sequence of runx2) effectively rescued the inhibition of BMSC osteogenic differentiation in miR-23b-overexpressing cells, indicating that the inhibiting effect of miR-23b on osteogenesis is mediated by suppression of runx2. Moreover, caudal vein injection of agomir-23b notably caused severe osteoporosis in mice, and forced expression of runx2 by combined injecting Ad-runx2 attenuated the bone loss induced by miR-23b. Collectively, these data indicated that miR-23b was involved in TNF-α-mediated reduction of BMSC osteogenesis by targeting runx2. These findings may provide new insights into understanding the regulatory role of miR-23b in the process of BMSC osteogenic differentiation in inflammatory conditions and a novel therapeutic target for osteoporosis.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , MicroRNAs/metabolismo , Osteogênese/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Regiões 3' não Traduzidas/genética , Animais , Sequência de Bases , Reabsorção Óssea/patologia , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Humanos , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
13.
Cell Death Dis ; 8(10): e3140, 2017 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-29072705

RESUMO

Cartilage dyshomeostasis contributes to osteoarthritis (OA) pathogenesis, and tumor necrosis factor (TNF)-α has critical role in this process by driving inflammatory cascades and cartilage degradation. However, the negative regulation of TNF-α-mediated signaling remains undefined. Here we demonstrate the crucial role of miR-145 in the modulation of TNF-α-mediated signaling and cartilage matrix degradation. MicroRNA (miRNA) expression profiles of TNF-α-stimulated chondrocytes showed that miR-145 expression was rapidly downregulated by TNF-α. Moreover, miR-145 was directly repressed by p65 and was negatively correlated with TNF-α secretion during OA progression. Further, we found that miR-145 directly targeted mitogen-activated protein kinase kinase 4 (MKK4) and broadly restrained the production of several TNF-α-triggered matrix-degrading enzymes (MMP-3, MMP-13, and Adamts-5). Mechanistic studies unveiled that miR-145 negatively regulated TNF-α-mediated JNK and p38 activation, as well as the nuclear accumulation of p-c-Jun and p-ATF2, by inhibiting MKK4 phosphorylation, eventually resulting in the alteration of catabolic genes transcription. Indeed, p-ATF2 interacted with the promoter of Mmp-13, whereas p-c-Jun bound to promoters of Mmp-3 and Adamts-5. MKK4 was significantly elevated in OA cartilage. Eliminating MKK4 by short hairpin RNA resulted in obviously decreased matrix-degrading enzymes production, JNK and p38 inactivation, and an inhibition of cartilage degradation. On the contrary, MKK4 overexpression enhanced TNF-α-mediated signaling activation and transcription of downstream catabolic genes, and consequently worsened cartilage degradation. Moreover, intra-articular (IA) injection of miR-145 agonist to rat with surgery-induced OA alleviated cartilage destruction. Altogether, we elucidate a novel regulatory mechanism underlying TNF-α-triggered cartilage degradation and demonstrate the potential utility of miR-145 and MKK4 as therapy targets for OA.


Assuntos
Cartilagem/metabolismo , MAP Quinase Quinase 4/metabolismo , MicroRNAs/metabolismo , Osteoartrite/genética , Fator de Necrose Tumoral alfa/metabolismo , Animais , Cartilagem/patologia , Regulação para Baixo , Humanos , Masculino , Osteoartrite/metabolismo , Osteoartrite/patologia , Ratos , Transfecção
14.
Front Pharmacol ; 8: 407, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28713268

RESUMO

Postmenopausal osteoporosis is a disease characterized by excessive osteoclastic bone resorption. Some anti-diabetic drugs were demonstrated for anti-osteoclastic bone-loss effects. The present study investigated the skeletal effects of chronic administration of sitagliptin, a dipeptidyl peptidase IV (DPP IV) inhibitor that is increasingly used for type 2 diabetes treatments, in an estrogen deficiency-induced osteoporosis and elucidated the associated mechanisms. This study indicated that sitagliptin effectively prevented ovariectomy-induced bone loss and reduced osteoclast numbers in vivo. It was also indicated that sitagliptin suppressed receptor activator of nuclear factor-κB ligand (RANKL)-mediated osteoclast differentiation, bone resorption, and F-actin ring formation in a manner of dose-dependence. In addition, sitagliptin significantly reduced the expression of osteoclast-specific markers in mouse bone-marrow-derived macrophages, including calcitonin receptor (Calcr), dendrite cell-specific transmembrane protein (Dc-stamp), c-Fos, and nuclear factor of activated T-cells cytoplasmic 1 (Nfatc1). Further study indicated that sitagliptin inhibited osteoclastogenesis by suppressing AKT and ERK signaling pathways, scavenging ROS activity, and suppressing the Ca2+ oscillation that consequently affects the expression and/or activity of the osteoclast-specific transcription factors, c-Fos and NFATc1. Collectively, these findings suggest that sitagliptin possesses beneficial effects on bone and the suppression of osteoclast number implies that the effect is exerted directly on osteoclastogenesis.

15.
Biol Open ; 6(8): 1130-1136, 2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28642243

RESUMO

Osteoarthritis (OA) is a common and dynamic disease of the joints, including the articular cartilage, underlying bones and synovium. In particular, OA is considered as the degeneration of the cartilage. Tectorigenin (Tec) is known to affect many biological processes; however, its effects on articular chondrocytes remain unclear. This study aimed to assess the effects of Tec on articular cartilage. In vitro, Tec inhibited the expression levels of type X collagen, cyclooxigenase-2, matrix metalloproteinase (MMP)-3 and MMP-13, but enhanced the expression of Runx1, type II collagen and aggrecan in the presence of IL-1ß. Meanwhile, Tec inhibited apoptosis through the Bax/Bcl-2/caspase-3 pathway, upregulating p-Bad, downregulating the Bax/Bcl-2 ratio, and activating caspase-3 compared with IL-1ß treatment only. Moreover, this process was partially regulated by NF-κB P65. In vivo, the chondroprotective effects of Tec were assessed by establishing a model of surgically induced OA. Tec-treated joints exhibited fewer osteoarthritic changes than saline-treated joints. Meanwhile, 1.5 µg/kg Tec treatment produced a greater protective effect than 0.75 µg/kg Tec. The Osteoarthritis Research Society International (OARSI) scoring system, employed to assess histopathological grading of the models, as well immunohistochemistry for Aggrecan Neoepitope and MMP-3, further confirmed the results. In conclusion, this study showed that Tec plays a chondroprotective role in the OA process by preventing articular cartilage degeneration and chondrocyte apoptosis via the NF-κB P65 pathway.

16.
Exp Cell Res ; 352(2): 346-356, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28215635

RESUMO

Mechanical unloading leads to bone loss and disuse osteoporosis partly due to impaired osteoblastogenesis of bone marrow stromal cells (BMSCs). However, the underlying molecular mechanisms of this phenomenon are not fully understood. In this study, we demonstrated that cyclic mechanical stretch (CMS) promotes osteoblastogenesis of BMSCs both in vivo and in vitro. Besides, we found that Hedgehog (Hh) signaling pathway was activated in this process. Inhibition of which by either knockdown of Sonic hedgehog (Shh) or treating BMSCs with Hh inhibitors attenuated the osteogenic effect of CMS on BMSCs, suggesting that Hh signaling pathway acts as an endogenous mediator of mechanical stimuli on BMSCs. Furthermore, we demonstrated that Shh expression level was regulated by DNA methylation mechanism. Chromatin Immunoprecipitation (ChIP) assay showed that DNA methyltransferase 3b (Dnmt3b) binds to Shh gene promoter, leading to DNA hypermethylation in mechanical unloading BMSCs. However, mechanical stimulation down-regulates the protein level of Dnmt3b, results in DNA demethylation and Shh expression. More importantly, we found that inhibition of Dnmt3b partly rescued bone loss in HU mice by mechanical unloading. Our results demonstrate, for the first time, that mechanical stimulation regulates osteoblastic genes expression via direct regulation of Dnmt3b, and the therapeutic inhibition of Dnmt3b may be an efficient strategy for enhancing bone formation under mechanical unloading.


Assuntos
Células da Medula Óssea/citologia , Epigênese Genética , Proteínas Hedgehog/genética , Células-Tronco Mesenquimais/citologia , Osteogênese , Estresse Mecânico , Animais , Células da Medula Óssea/metabolismo , Células Cultivadas , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Proteínas Hedgehog/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/citologia , Osteoblastos/metabolismo , Ligação Proteica , DNA Metiltransferase 3B
17.
Biochem Biophys Res Commun ; 485(2): 400-408, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28213129

RESUMO

Developmental dysplasia of the hip (DDH) is a common musculoskeletal disorder characterized by a mismatch between acetabulum and femoral head. Mechanical force plays an important role during the occurrence and development of abnormities in acetabulum and femoral head. In this study, we established a mechanical force model named cyclic compressive stress (Ccs). To analyze the effect of Ccs on DDH, we detected special genes in chondrocytes and osteoblasts. Results showed that Ccs downregulated chondrogenesis of ADTC5 in a concentration-dependent manner. Moreover, the mRNA level of Scinderin (Scin) considerably increased. We established lentivirus-SCIN(GV144-SCIN) to transfect hBMSCs, which were treated with different Ccs levels (0.25 Hz*5 cm, 0.5 Hz*5 cm, and 1 Hz*10 cm); the result showed that overexpression of Scin upregulated osteogenesis and osteoclastogenesis. By contrast, expression of chondrocyte-specific genes, including ACAN, COL-2A, and Sox9, decreased. Further molecular investigation demonstrated that Scin promoted osteogenesis and osteoclastogenesis through activation of the p-Smad1/5/8, NF-κB, and MAPK P38 signaling pathways, as well as stimulated the expression of key osteoclast transcriptional factors NFATc1 and c-Fos. Moreover, Scin-induced osteogenesis outweighed osteoclastogenesis in defective femur in vivo. The results of the analysis of Micro-CT confirmed these findings. Overall, Ccs influenced the development of DDH by promoting osteogenesis and cartilage degradation. In addition, Scin played a vital role in the development of DDH.


Assuntos
Gelsolina/genética , Regulação da Expressão Gênica , Luxação Congênita de Quadril/genética , Estresse Mecânico , Animais , Western Blotting , Linhagem Celular Tumoral , Células Cultivadas , Condrócitos/metabolismo , Condrogênese/genética , Progressão da Doença , Gelsolina/metabolismo , Luxação Congênita de Quadril/metabolismo , Luxação Congênita de Quadril/patologia , Humanos , Sistema de Sinalização das MAP Quinases , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos Nus , NF-kappa B/metabolismo , Osteoblastos/metabolismo , Osteogênese/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo
18.
J Cell Biochem ; 118(2): 286-297, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27335248

RESUMO

Osteoblasts are essential for maintaining skeletal architecture and modulating bone microenvironment homeostasis. From numerous associated investigations, the BMP-2 pathway has been well-defined as a vital positive modulator of bone homeostasis. Gremlin2 (Grem2) is a bone morphogenetic protein (BMP) antagonists. However, the effect of Grem2 on the BMP-2-induced osteogenesis of human bone marrow-derived mesenchymal stem cells (hBMSCs) remains ambiguous. This study aimed to analyze the procedure in vitro and in vivo. The differentiation of hBMSCs was assessed by determining the expression levels of several osteoblastic genes, as well as the enzymatic activity and calcification of alkaline phosphatase. We found that Grem2 expression was upregulated by BMP-2 within the range of 0-1 µg/mL, and significant increases were evident at 48, 72, and 96 h after BMP-2 treatment. Si-Grem2 increased the BMP-2-induced osteogenic differentiation of hBMSCs, whereas overexpression of Grem2 had the opposite trend. The result was confirmed using a defective femur model. We also discovered that the BMP-2/Smad/Runx2 pathway played an important role in the process. This study showed that si-Grem2 increased the BMP-2-induced osteogenic differentiation of hBMSCs via the BMP-2/Smad/Runx2 pathway. J. Cell. Biochem. 118: 286-297, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Células da Medula Óssea/metabolismo , Proteína Morfogenética Óssea 2/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Transdução de Sinais , Proteínas Smad/metabolismo , Células da Medula Óssea/citologia , Proteína Morfogenética Óssea 2/genética , Diferenciação Celular , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Citocinas , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Células-Tronco Mesenquimais/citologia , Proteínas Smad/genética
19.
Cell Death Dis ; 7(8): e2335, 2016 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-27512956

RESUMO

Bone morphogenetic protein 2 (BMP2) has been used to induce bone regeneration by promoting osteogenic differentiation of bone marrow-derived mesenchymal stem cells (MSCs). However, its effect is attenuated in osteoporotic conditions by unknown mechanisms. In this study, we investigated the molecular mechanisms of reduced osteogenic effect of BMP2 in osteoporotic conditions. By interrogating the microarray data from osteoporosis patients, we revealed an upregulation of the epigenetic modifying protein lysine (K)-specific demethylase 5A (KDM5A) and decreased Runt-related transcription factor 2 (RUNX2) expression. Further studies were focused on the role of KDM5A in osteoporosis. We first established ovariectomized (OVX) mouse model and found that the BMP2-induced osteogenic differentiation of osteoporotic MSCs was impaired. The elevated level of KDM5A was confirmed in osteoporotic MSCs. Overexpression of KDM5A in normal MSCs inhibited BMP2-induced osteogenesis. Moreover, osteogenic differentiation of osteoporotic MSCs was restored by specific KDM5A short hairpin RNA or inhibitor. Furthermore, by chromatin immunoprecipitation assay we demonstrated that KDM5A functions as endogenous modulator of osteogenic differentiation by decreasing H3K4me3 levels on promoters of Runx2, depend on its histone methylation activity. More importantly, we found an inhibitory role of KDM5A in regulating bone formation in osteoporotic mice, and pretreatment with KDM5A inhibitor partly rescued the bone loss during osteoporosis. Our results show, for the first time, that KDM5A-mediated H3K4me3 modification participated in the etiology of osteoporosis and may provide new strategies to improve the clinical efficacy of BMP2 in osteoporotic conditions.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Osso e Ossos/patologia , Diferenciação Celular , Células-Tronco Mesenquimais/patologia , Osteogênese , Osteoporose/patologia , Proteína 2 de Ligação ao Retinoblastoma/metabolismo , Idoso , Animais , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Histonas/metabolismo , Humanos , Lisina/metabolismo , Células-Tronco Mesenquimais/metabolismo , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Osteoporose/metabolismo , Ovariectomia , Regiões Promotoras Genéticas/genética
20.
Dalton Trans ; 44(40): 17774-83, 2015 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-26399730

RESUMO

Two Zn(ii) and Cd(ii) coordination polymers [ZnL2·2DMF] (1) and [CdL(OAc)] (2) were first synthesized by treating a novel 2-substituted 8-hydroxyquinolinate ligand HL involving a pyridyl group with zinc or cadmium salts. Two dicarboxylic acid ligands (H2BDC = 1,4-benzenedicarboxylic acid; H2BPDC = 4,4'-biphenyldicarboxylic acid) are employed as secondary auxiliary ligands to perform a systematic study on the structural diversities in the M(ii)-quinolinate frameworks. By introducing two dicarboxylate anions in the reaction system, four new polymers [Zn2L2(BDC)] (3), [Zn3L2I2(BPDC)·2MeOH·8H2O] (4), [Cd2L2(BDC)] (5) and [Cd2L2(BPDC)·2MeOH·4H2O] (6) were obtained. Complex 1 possesses a two-dimensional (2D) square grid containing meso-helical chains. Complex 2 is a 2D network fabricated by binuclear {Zn2} secondary building units (SBUs). Complexes 3 and 5 show a kind of 2D structure constructed by cyclic hexamers Zn6L4, which are divided into half by the coordinated BDC. In complex 4, the BPDC ligands bridge the 1D M(ii)-L chains into a 2D layered structure. Complex 6 presents an interesting 3D structure, in which the BPDC ligands link the binuclear Cd(ii) units into many meso-helical chains along the a and b axes. The diverse structures of complexes 1-6 indicate that the skeletons of dicarboxylate anions play an important role in the assembly of such different frameworks. Moreover, distinct fluorescence properties (emission wavelength and lifetime) of the complexes 1-6 were observed in the solid state.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...